Detection of Normal and Chimeric Nucleophosmin in Human Cells

Blood ◽  
1999 ◽  
Vol 93 (2) ◽  
pp. 632-642 ◽  
Author(s):  
Jacqueline L. Cordell ◽  
Karen A.F. Pulford ◽  
Barbara Bigerna ◽  
Giovanna Roncador ◽  
Alison Banham ◽  
...  

In anaplastic large-cell lymphoma (ALCL), the (2;5) chromosomal translocation creates a fusion gene encoding the 80-kD NPM-ALK hybrid protein. This report describes three new monoclonal antibodies, two of which recognize, by Western blotting, the N-terminal portion of NPM present in the NPM-ALK fusion protein and also in two other NPM fusion proteins (NPM-RAR and NPM-MLF1). The third antibody recognizes the C-terminal portion (deleted in NPM-ALK) and reacts only with wild-type NPM. The three antibodies immunostain wild-type NPM (in paraffin-embedded normal tissue samples) in cell nuclei and in the cytoplasm of mitotic cells. Cerebral neurones, exceptionally, show diffuse cytoplasmic labeling. In contrast to normal tissues, the two antibodies against the N-terminal portion of NPM labeled the cytoplasm of neoplastic cells, in four ALK-positive ALCL, reflecting their reactivity with NPM-ALK fusion protein, whereas the antibody to the C-terminal NPM epitope labeled only cell nuclei. Immunocytochemical labeling with these antibodies can therefore confirm that an ALK-positive lymphoma expresses NPM-ALK (rather than a variant ALK-fusion protein) and may also provide evidence for chromosomal anomalies involving the NPM gene other than the classical (2;5) translocation.

Blood ◽  
1999 ◽  
Vol 93 (2) ◽  
pp. 632-642 ◽  
Author(s):  
Jacqueline L. Cordell ◽  
Karen A.F. Pulford ◽  
Barbara Bigerna ◽  
Giovanna Roncador ◽  
Alison Banham ◽  
...  

Abstract In anaplastic large-cell lymphoma (ALCL), the (2;5) chromosomal translocation creates a fusion gene encoding the 80-kD NPM-ALK hybrid protein. This report describes three new monoclonal antibodies, two of which recognize, by Western blotting, the N-terminal portion of NPM present in the NPM-ALK fusion protein and also in two other NPM fusion proteins (NPM-RAR and NPM-MLF1). The third antibody recognizes the C-terminal portion (deleted in NPM-ALK) and reacts only with wild-type NPM. The three antibodies immunostain wild-type NPM (in paraffin-embedded normal tissue samples) in cell nuclei and in the cytoplasm of mitotic cells. Cerebral neurones, exceptionally, show diffuse cytoplasmic labeling. In contrast to normal tissues, the two antibodies against the N-terminal portion of NPM labeled the cytoplasm of neoplastic cells, in four ALK-positive ALCL, reflecting their reactivity with NPM-ALK fusion protein, whereas the antibody to the C-terminal NPM epitope labeled only cell nuclei. Immunocytochemical labeling with these antibodies can therefore confirm that an ALK-positive lymphoma expresses NPM-ALK (rather than a variant ALK-fusion protein) and may also provide evidence for chromosomal anomalies involving the NPM gene other than the classical (2;5) translocation.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 460-460 ◽  
Author(s):  
Charlotte Gruber ◽  
Rebekka Grundler ◽  
Georg Haecker ◽  
Falco Fend ◽  
Christian Peschel ◽  
...  

Abstract Anaplastic large cell lymphomas (ALCLs) define a subgroup of aggressive non-Hodgkin’s lymphomas. In 40–60% of systemic ALCLs, a t(2;5) (p23;q35) translocation is found, which generates a fusion gene between nucleophosmin (NPM) and the receptor tyrosine kinase gene ALK (anaplastic lymphoma kinase). The NPM-ALK chimeric gene encodes a constitutively activated tyrosine kinase and is believed to initiate the process of lymphomagenesis. Since NPM-ALK has been shown to activate PI3-kinase and STAT3, proteins that are involved in apoptosis regulation, altered apoptosis might contribute to ALCL development. Bcl-3, which exerts an anti-apoptotic effect in B and T lymphocytes, has been reported to be up regulated in a subgroup of ALCLs. This raised the question whether Bcl-3 is required for tumour development induced by NPM-ALK. In a first set of experiments, the expression levels of Bcl-3 in NPM-ALK negative and NPM-ALK positive tumour cells were analysed. Bcl-3 expression was enhanced in NPM-ALK expressing cells compared to NPM-ALK negative cells or cells expressing a kinase dead NPM-ALK mutant. Next we used primary murine bone marrow cells from wt versus Bcl-3 −/− mice in order to clarify the contribution of Bcl-3 to NPM-ALK induced transformation. We employed a retroviral infection system utilizing a MCSV-based vector co-expressing NPM-ALK together with the enhanced green fluorescent protein (EGFP) via an internal ribosomal entry site to infect Bcl-3−/− and wild type (wt) bone marrow cells. Transformation of bone marrow cells was analysed by methylcellulose assay without cytokines. Transformation of NPM-ALK infected Bcl-3−/− bone marrow cells was comparable to NPM-ALK infected BL6 wt bone marrow cells. No colony formation was detectable after transfection of Bcl-3−/− bone marrow with empty vector as control. Finally, we utilized a murine transplantation model of ALCL. Lethally irradiated BL6 wt mice were transplanted with retrovirally NPM-ALK infected wild type or Bcl-3−/− bone marrow cells. As a control, BL6 wt mice received a transplant of Bcl-3−/− bone marrow cells infected with supernatant from viral producer cells transfected with empty vector. Mice transplanted with NPM-ALK infected wild type or Bcl-3−/− bone marrow cells rapidly died within a median survival time of 16 and 17 days respectively, whereas mice transplanted with Bcl-3 −/− bone marrow cells transfected with empty vector survived healthy for more than 300 days. Diseased mice macroscopically showed involvement of the spleen, predominantly. Histologically, spleens of diseased mice showed an extensive infiltration of ALK-positive tumour cells with proliferation of large histiocytic cells both in mice transplanted with NPM-ALK infected Bcl-3−/− and wild type bone marrow. In both groups, FACS-analysis revealed a high percentage of EGFP positive and thus NPM-ALK positive cells in bone marrow and spleen. In conclusion, NPM-ALK is able to transform bone marrow cells and to induce a lymphoma-like disease in the absence of Bcl-3. Bcl-3 is thus dispensable for ALCL development in a murine mouse model. Lack of Bcl-3 in the knockout mice may be compensated by the expression of other proteins. Therefore, Bcl-3 upregulation in a subgroup of human ALCLs may be not critical for lymphoma development.


Blood ◽  
1998 ◽  
Vol 92 (8) ◽  
pp. 2688-2695 ◽  
Author(s):  
Iwona Wlodarska ◽  
Chris De Wolf-Peeters ◽  
Brunangelo Falini ◽  
Gregor Verhoef ◽  
Stephan W. Morris ◽  
...  

Recently, a distinctive entity characterized by expression of the anaplastic lymphoma kinase (ALK) protein [most frequently due to the t(2;5)(p23;q35)-associated NPM-ALK fusion] has emerged within the heterogenous group of non-Hodgkin’s lymphomas (NHL) classified as anaplastic large-cell lymphoma (ALCL). Sporadic variant 2p23/ALK abnormalities identified in ALK-positive ALCL indicate that genes other than NPM may also be involved in the deregulation of ALK and lymphomagenesis. We report here three cases with an inv(2)(p23q35) detected by fluorescence in situ hybridization (FISH) in young male patients with ALK-positive ALCL. In contrast to ALCL cases with the classical t(2;5)(p23;q35) that usually show both cytoplasmic and nuclear or predominantly nuclear alone localization of the NPM-ALK chimeric product, in all three cases with an inv(2)(p23q35) the ALK protein accumulated in the cytoplasm only, supporting the previous assumption that the oncogenic potential of ALK may not be dependent on its nuclear localization. As the first step to identify theALK partner gene involved in the inv(2)(p23q35), we performed extensive FISH studies and demonstrated that the 2q35 breakpoint occurred within the 1,750-kb region contained within the 914E7 YAC. Moreover, a striking association of the inv(2)(p23q35) with a secondary chromosomal change, viz, ider(2)(q10)inv(2)(p23q35), carrying two additional copies of the putative ALK-related fusion gene, was found in all three patients, suggesting that, in contrast to the standard t(2;5)/NPM-ALK fusion, multiple copies of the putative 2q35-ALK chimeric gene may be required for efficient tumor development. In summary, we demonstrate that the inv(2)(p23q35), a variant of the t(2;5)(p23;q35), is a recurrent chromosomal abnormality in ALK-positive ALCL, the further characterization of which should provide new insight into the pathogenesis of these lymphomas. © 1998 by The American Society of Hematology.


2015 ◽  
Vol 112 (11) ◽  
pp. E1272-E1277 ◽  
Author(s):  
Kalpana Kannan ◽  
Cristian Coarfa ◽  
Pei-Wen Chao ◽  
Liming Luo ◽  
Yan Wang ◽  
...  

High-grade serous ovarian cancer (HGSC) is among the most lethal forms of cancer in women. Excessive genomic rearrangements, which are expected to create fusion oncogenes, are the hallmark of this cancer. Here we report a cancer-specific gene fusion between BCAM, a membrane adhesion molecule, and AKT2, a key kinase in the PI3K signaling pathway. This fusion is present in 7% of the 60 patient cancers tested, a significant frequency considering the highly heterogeneous nature of this malignancy. Further, we provide direct evidence that BCAM-AKT2 is translated into an in-frame fusion protein in the patient’s tumor. The resulting AKT2 fusion kinase is membrane-associated, constitutively phosphorylated, and activated as a functional kinase in cells. Unlike endogenous AKT2, whose activity is tightly regulated by external stimuli, BCAM-AKT2 escapes the regulation from external stimuli. Moreover, a BCAM-AKT2 fusion gene generated via chromosomal translocation using the CRISPR/Cas9 system leads to focus formation in both OVCAR8 and HEK-293T cell lines, suggesting that BCAM-AKT2 is oncogenic. Together, the results indicate that BCAM-AKT2 expression is a new mechanism of AKT2 kinase activation in HGSC. BCAM-AKT2 is the only fusion gene in HGSC that is proven to translate an aberrant yet functional kinase fusion protein with oncogenic properties. This recurrent genomic alteration is a potential therapeutic target and marker of a clinically relevant subtype for tailored therapy of HGSC.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3571-3571
Author(s):  
Claudia Fuchs ◽  
Paul Vesely ◽  
Isabella Bambach ◽  
Silvia Schauer ◽  
Werner Linkesch ◽  
...  

Abstract Anaplastic large cell lymphoma (ALCL) accounts for approximately 30% of childhood lymphomas and 3% of adult non-Hodgkin lymphomas. The nucleophosmin - anaplastic lymphoma kinase (NPM-ALK) fusion which is the product of a t(2;5)(p23;q35) chromosomal translocation is present in about half of nodal ALCL. Expression of this fusion kinase results in induction of the AP-1 transcription factor JunB and IL-3 independent outgrow of murine hematopoietic Ba/F3 cells. We demonstrated that wild type NPM-ALK increases the amount of ribosomes bound to JUNB mRNA resulting in its more effective translation in large polysomes. The NPM-ALK fusion tyrosine kinase has 20 potential tyrosine residues available for autophosphorylation and phosphorylation by other protein tyrosine kinases. Here we used series of Y-to-F-substituted mutants of NPM-ALK to identify tyrosine residues that are required to regulate the segregation of JUNB mRNAs between polysomes and monosomes as well as ribonucleic particles (RNPs). Neither JUNB transcription nor JunB translation was altered in Ba/F3 cells expressing NPM-ALK mutants Y17F/Y29F/Y67F Y138F/Y152F Y156F/Y191F/Y299F Y378F/Y418F/Y445F and Y646F/Y664F compared to NPM-ALK wild type. Conversely, in NPM-ALK Y567F/Y461F/Y644F mutant cells proliferation was markedly decreased. These cells demonstrated active MEK-ERK pathway, while AKT, mTOR, and rpS6 phosphorylation was impaired. Moreover a shift of JUNB mRNA from the polysomic to the monosomic/mRNP fraction could be observed. In conclusion, we identified specific NPM-ALK phosphorylation sites required to mediate the effect of NPM-ALK on the JUNB translational regulation and therefore provide further insights in the transforming mechanisms of the oncoprotein NPM-ALK.


Blood ◽  
1998 ◽  
Vol 92 (8) ◽  
pp. 2688-2695 ◽  
Author(s):  
Iwona Wlodarska ◽  
Chris De Wolf-Peeters ◽  
Brunangelo Falini ◽  
Gregor Verhoef ◽  
Stephan W. Morris ◽  
...  

Abstract Recently, a distinctive entity characterized by expression of the anaplastic lymphoma kinase (ALK) protein [most frequently due to the t(2;5)(p23;q35)-associated NPM-ALK fusion] has emerged within the heterogenous group of non-Hodgkin’s lymphomas (NHL) classified as anaplastic large-cell lymphoma (ALCL). Sporadic variant 2p23/ALK abnormalities identified in ALK-positive ALCL indicate that genes other than NPM may also be involved in the deregulation of ALK and lymphomagenesis. We report here three cases with an inv(2)(p23q35) detected by fluorescence in situ hybridization (FISH) in young male patients with ALK-positive ALCL. In contrast to ALCL cases with the classical t(2;5)(p23;q35) that usually show both cytoplasmic and nuclear or predominantly nuclear alone localization of the NPM-ALK chimeric product, in all three cases with an inv(2)(p23q35) the ALK protein accumulated in the cytoplasm only, supporting the previous assumption that the oncogenic potential of ALK may not be dependent on its nuclear localization. As the first step to identify theALK partner gene involved in the inv(2)(p23q35), we performed extensive FISH studies and demonstrated that the 2q35 breakpoint occurred within the 1,750-kb region contained within the 914E7 YAC. Moreover, a striking association of the inv(2)(p23q35) with a secondary chromosomal change, viz, ider(2)(q10)inv(2)(p23q35), carrying two additional copies of the putative ALK-related fusion gene, was found in all three patients, suggesting that, in contrast to the standard t(2;5)/NPM-ALK fusion, multiple copies of the putative 2q35-ALK chimeric gene may be required for efficient tumor development. In summary, we demonstrate that the inv(2)(p23q35), a variant of the t(2;5)(p23;q35), is a recurrent chromosomal abnormality in ALK-positive ALCL, the further characterization of which should provide new insight into the pathogenesis of these lymphomas. © 1998 by The American Society of Hematology.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 27-28
Author(s):  
Xiaoqian Liu ◽  
Yuping Li ◽  
Xuxiang Liu ◽  
Wei Qi ◽  
Jibin Zhang ◽  
...  

Recurrent VAV1 mutations and gene fusions (VAV1-THAP4, VAV1-MYO1F, and VAV1-S100A7) have been identified in peripheral T-cell lymphoma (PTCL) including angioimmunoblastic T-cell lymphoma (AITL) patients. A common theme of these genetic aberrations is the loss of the auto-inhibitory C-terminal SH3 domain of VAV1 resulting in aberrant activation of VAV1 independent of normal activation events. Although mouse models support VAV1 mutation/fusion as having a driver oncogenic role in the pathogenesis of PTCL, investigations on VAV1 activity in human cells were performed mainly on the Jurkat cell line with exogenous expression of VAV1 fusion proteins. This approach has un-physiological expression of VAV1 and the functions of VAV1 fusion/mutation under normal endogenous regulation need to be explored. In this study, we introduced a fusion gene, similar to what has been observed in PTCL, into the endogenous VAV1 locus. The fusion gene was under normal regulatory controls instead of being over-expressed by a viral vector, thus providing a more accurate assessment of its function in vivo. To simulate VAV1 fusion, we knocked in a green fluorescence protein (GFP) sequence followed by a simian virus 40 (SV40) poly(A) signal into intron 25 of VAV1 locus by clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 (Cas9) technology. A homologous DNA repair (HDR) template with tandem homologous sequences of VAV1 gene, GFP gene, and a SV40 transcription poly(A) signal was electroporated into Jurkat cells together with the Cas9/sgRNA ribonucleoprotein (RNP) complex. This knock-in disrupted the transcription of exon 26 and exon 27, resulting in an in-frame fusion protein with GFP fused to the C-terminal of SH2 of VAV1 (VAV1SH2-GFP)(Figure A). Because our guide RNA targeted the intron 25 sequence by CRISPR/Cas9 system, any possible indels caused by non-homologous end joining will occur within the intron and will not change the protein sequence of the wild type VAV1. The GFP expressing cells were isolated from the edited cell population by FACS. The fusion of GFP with VAV1 in the sorted cells was confirmed by western blot (Figure B) and these cells displayed a heterozygous VAV1SH2-GFP fusion/wild type (WT) phenotype that mimicked the VAV1 translocations observed in PTCL patients. Jurkat cells with VAV1SH2-GFP showed spontaneous activation of the T-cell receptor (TCR) signaling pathway. Analysis of signaling events downstream of VAV1 demonstrated increased phosphorylation in ITK, LCK, and subsequent ERK in Jurkat cells with VAV1SH2-GFP compared with WT Jurkat cells by western blot (Figure B). We also observed consistently elevated pERK in Jurkat cells with VAV1SH2-GFP by flow cytometry (Figure C). Notably, this elevation in pERK was spontaneous and independent of TCR stimulation with anti-CD3 antibody. VAV1SH2-GFP fusion protein also led to marked activation of downstream NFAT and NF-κB pathways as shown by Luciferase reporter assays (Figure D). Similarly, the enhanced NFAT and NF-κB pathway activation in Jurkat with the fusion protein was independent of TCR stimulation. Interestingly, with antiCD3 stimulation, Jurkat cells with VAV1SH2-GFP showed significantly lower pERK, NFAT and NF-κB activity compared to WT Jurkat cells with anti-CD3 stimulation. In conclusion, in Jurkat cells genetically edited with VAV1SH2-GFP, spontaneous activation of TCR signaling and subsequently increased NFAT and NF-κB activity were observed. Our findings further support that VAV1 C-terminal SH3 domain plays an important regulatory role in blocking VAV1 activity in the absence of proper activation. Removing C-terminal SH3 domain or replacing it with GFP or other protein relieves this inhibition, allowing spontaneous activation independent of TCR stimulation. Our study also indicates the sensitivity of the TCR signaling pathway to the level of activation and hyperactivation is detrimental. To validate the function of VAV1SH2-GFP in normal T cells, we have also successfully edited primary CD4+ T cells with VAV1SH2-GFP and the TCR signaling pathway in edited primary CD4+ T cell is currently being evaluated with and without anti-CD3/CD28 stimulation. Figure Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2000 ◽  
Vol 95 (10) ◽  
pp. 3204-3207 ◽  
Author(s):  
Christian Touriol ◽  
Catherine Greenland ◽  
Laurence Lamant ◽  
Karen Pulford ◽  
Frédéric Bernard ◽  
...  

Abstract Anaplastic lymphoma kinase (ALK)-positive lymphomas are characterized by expression of a hybrid protein, comprising the cytoplasmic portion of the ALK tyrosine kinase fused to a partner protein. This hybrid kinase is often encoded by the nucleophosmin (NPM)NPM-ALK fusion gene resulting from the (2;5)(p23;q35) chromosomal translocation. However, the ALK gene at 2p23 may also be involved in 2 variant translocations, namely t(1;2)(q25;p23) and t(2;3)(p23;q21), which create the TPM3-ALK andTFG-ALK fusion genes, respectively. We report here 2 lymphomas with an unusual finely granular cytoplasmic ALK staining pattern, clearly different from the pattern observed in ALK-positive lymphomas carrying NPM-ALK or its variants. A cloned complementary DNA sequence from 1 of these 2 lymphomas contained the ALK gene fused to the second clathrin heavy chain gene (also referred to as clathrin heavy polypeptide-like gene) (CLTCL). The distinctive granular cytoplasmic staining pattern for ALK was likely to be due to binding of the fusion protein to clathrin-coated vesicles. TheCLTCL gene is constitutively expressed in lymphoid cells and therefore presumably contributes an active promoter for theCLTCL-ALK gene. The fusion protein had a molecular weight (250 kd) that differs from all known ALK products, and it was autophosphorylated in an in vitro kinase assay, confirming that it is constitutively active and hence capable of contributing to malignant transformation. These 2 cases, therefore, represent a hitherto undescribed mechanism of ALK activation in lymphoma and further illustrate the diversity of fusion partners for the ALKgene.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4331-4331
Author(s):  
Roberto Tonelli ◽  
Luca Lo Nigro ◽  
Salvatore Serravalle ◽  
Luca Montemurro ◽  
Giancarlo Izzi ◽  
...  

Abstract Rearrangement 11q23, involving MLL gene is mainly associated with M4/M5 acute myeloid leukemia (AML) and is characterized by poor prognosis and responsiveness to therapy. In this case blasts from an infant patient with monocytic AML were analyzed at diagnosis. The most frequent translocations of MLL, t(4;11) (40%, MLL-AF4) and t(9;11) (27%, MLL-AF9) were investigated by RT-PCR and resulted negative, meanwhile interphase FISH analysis, performed by using specific MLL probes, revealed a MLL gene translocation. Panhandle-PCR technique was performed to identify the partner gene and candidate clones DNA were sequenced to identify the unknown partner gene. The sequence analysis revealed the presence of a region of MLL connected with a region of ArgBP2 gene (chromosome band 4q35.1). The fusion gene is composed of the first 9 MLL exons and the last 3 ArgBP2 exons (exons 19–21) (Fig. 1). ArgBP2 belongs to vinexines family of protein adapters with three C-terminal SH3 domains, several potential phosphorilation sites for c-Abl and a serine/threonine rich region. The predicted fusion protein consists of AT-hooks and MT domains of MLL and the last ArgBP2 SH3 domain. MLL-ArgBP2 chimeric transcript was expressed in the blast while the reciprocal transcript ArgBP2-MLL was not. Identification of this new translocation of MLL gene confirms the high grade of MLL recombination in infant AMLs. Ongoing studies are in progress to analyze the mechanism leading to this new chromosomal translocation and the leukemogenic potentialities of the new MLL-ArgBP2 fusion protein. Figure Figure


Sign in / Sign up

Export Citation Format

Share Document